Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood ; 138(23): 2408-2424, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34324649

RESUMO

Immune thrombocytopenia (ITP) is a platelet disorder. Pediatric and adult ITP have been associated with sialic acid alterations, but the pathophysiology of ITP remains elusive, and ITP is often a diagnosis of exclusion. Our analysis of pediatric ITP plasma samples showed increased anti-Thomsen-Friedenreich antigen (TF antigen) antibody representation, suggesting increased exposure of the typically sialylated and cryptic TF antigen in these patients. The O-glycan sialyltransferase St3gal1 adds sialic acid specifically on the TF antigen. To understand if TF antigen exposure associates with thrombocytopenia, we generated a mouse model with targeted deletion of St3gal1 in megakaryocytes (MK) (St3gal1MK-/-). TF antigen exposure was restricted to MKs and resulted in thrombocytopenia. Deletion of Jak3 in St3gal1MK-/- mice normalized platelet counts implicating involvement of immune cells. Interferon-producing Siglec H-positive bone marrow (BM) immune cells engaged with O-glycan sialic acid moieties to regulate type I interferon secretion and platelet release (thrombopoiesis), as evidenced by partially normalized platelet count following inhibition of interferon and Siglec H receptors. Single-cell RNA-sequencing determined that TF antigen exposure by MKs primed St3gal1MK-/- BM immune cells to release type I interferon. Single-cell RNA-sequencing further revealed a new population of immune cells with a plasmacytoid dendritic cell-like signature and concomitant upregulation of the immunoglobulin rearrangement gene transcripts Igkc and Ighm, suggesting additional immune regulatory mechanisms. Thus, aberrant TF antigen moieties, often found in pathological conditions, regulate immune cells and thrombopoiesis in the BM, leading to reduced platelet count.


Assuntos
Megacariócitos/patologia , Contagem de Plaquetas , Polissacarídeos/análise , Púrpura Trombocitopênica Idiopática/patologia , Adolescente , Animais , Antígenos Glicosídicos Associados a Tumores/análise , Criança , Pré-Escolar , Humanos , Lactente , Camundongos Endogâmicos C57BL , Sialiltransferases/análise , beta-Galactosídeo alfa-2,3-Sialiltransferase
2.
Blood ; 137(15): 2085-2089, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33238000

RESUMO

Aberrant megakaryopoiesis is a hallmark of the myeloproliferative neoplasms (MPNs), a group of clonal hematological malignancies originating from hematopoietic stem cells, leading to an increase in mature blood cells in the peripheral blood. Sialylated derivatives of the glycan structure ß4-N-acetyllactosamine (Galß1,4GlcNAc or type-2 LacNAc, hereafter referred to as LacNAc) regulate platelet life span, hepatic thrombopoietin (TPO) production, and thrombopoiesis. We found increased TPO plasma levels in MPNs with high allele burden of the mutated clones. Remarkably, platelets isolated from MPNs had a significant increase in LacNAc expression that correlated with the high allele burden regardless of the underlying identified mutation. Megakaryocytes derived in vitro from these patients showed an increased expression of the B4GALT1 gene encoding ß-1,4-galactosyltransferase 1 (ß4GalT1). Consistently, megakaryocytes from MPN showed increased LacNAc expression relative to healthy controls, which was counteracted by the treatment with a Janus kinase 1/2 inhibitor. Altered expression of B4GALT1 in mutant megakaryocytes can lead to the production of platelets with aberrant galactosylation, which in turn promote hepatic TPO synthesis regardless of platelet mass. Our findings provide a new paradigm for understanding aberrant megakaryopoiesis in MPNs and identify ß4GalT1 as a potential actionable target for therapy.


Assuntos
Plaquetas/patologia , Galactose/metabolismo , Galactosiltransferases/genética , Transtornos Mieloproliferativos/genética , Trombopoetina/sangue , Plaquetas/metabolismo , Galactose/análise , Galactosiltransferases/metabolismo , Humanos , Megacariócitos/metabolismo , Megacariócitos/patologia , Mutação , Transtornos Mieloproliferativos/sangue , Transtornos Mieloproliferativos/metabolismo , Trombopoetina/metabolismo , Regulação para Cima
3.
Nat Commun ; 11(1): 356, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31953383

RESUMO

Glycosylation is critical to megakaryocyte (MK) and thrombopoiesis in the context of gene mutations that affect sialylation and galactosylation. Here, we identify the conserved B4galt1 gene as a critical regulator of thrombopoiesis in MKs. ß4GalT1 deficiency increases the number of fully differentiated MKs. However, the resulting lack of glycosylation enhances ß1 integrin signaling leading to dysplastic MKs with severely impaired demarcation system formation and thrombopoiesis. Platelets lacking ß4GalT1 adhere avidly to ß1 integrin ligands laminin, fibronectin, and collagen, while other platelet functions are normal. Impaired thrombopoiesis leads to increased plasma thrombopoietin (TPO) levels and perturbed hematopoietic stem cells (HSCs). Remarkably, ß1 integrin deletion, specifically in MKs, restores thrombopoiesis. TPO and CXCL12 regulate ß4GalT1 in the MK lineage. Thus, our findings establish a non-redundant role for ß4GalT1 in the regulation of ß1 integrin function and signaling during thrombopoiesis. Defective thrombopoiesis and lack of ß4GalT1 further affect HSC homeostasis.


Assuntos
Galactosiltransferases/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Homeostase , Integrina beta1/metabolismo , Trombopoese/fisiologia , Animais , Plaquetas/metabolismo , Plaquetas/patologia , Adesão Celular , Diferenciação Celular , Quimiocina CXCL12/metabolismo , Colágeno , Modelos Animais de Doenças , Fibronectinas , Galactosiltransferases/genética , Predisposição Genética para Doença , Hemorragia/genética , Hemorragia/metabolismo , Hemorragia/patologia , Integrina beta1/genética , Laminina , Ligantes , Megacariócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patologia , Trombopoese/genética , Trombopoetina/sangue
4.
Cancer Discov ; 6(4): 368-81, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26951227

RESUMO

UNLABELLED: Somatic mutations in calreticulin (CALR) are present in approximately 40% of patients with myeloproliferative neoplasms (MPN), but the mechanism by which mutant CALR is oncogenic remains unclear. Here, we demonstrate that expression of mutant CALR alone is sufficient to engender MPN in mice and recapitulates the disease phenotype of patients with CALR-mutant MPN. We further show that the thrombopoietin receptor MPL is required for mutant CALR-driven transformation through JAK-STAT pathway activation, thus rendering mutant CALR-transformed hematopoietic cells sensitive to JAK2 inhibition. Finally, we demonstrate that the oncogenicity of mutant CALR is dependent on the positive electrostatic charge of the C-terminus of the mutant protein, which is necessary for physical interaction between mutant CALR and MPL. Together, our findings elucidate a novel paradigm of cancer pathogenesis and reveal how CALR mutations induce MPN. SIGNIFICANCE: The mechanism by which CALR mutations induce MPN remains unknown. In this report, we show that the positive charge of the CALR mutant C-terminus is necessary to transform hematopoietic cells by enabling binding between mutant CALR and the thrombopoietin receptor MPL.


Assuntos
Calreticulina/genética , Transformação Celular Neoplásica/genética , Mutação , Domínios e Motivos de Interação entre Proteínas/genética , Receptores de Trombopoetina/genética , Animais , Sequência de Bases , Transplante de Medula Óssea , Calreticulina/química , Calreticulina/metabolismo , Linhagem Celular , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Feminino , Mutação da Fase de Leitura , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Janus Quinases/antagonistas & inibidores , Janus Quinases/metabolismo , Camundongos , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Fenótipo , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , Receptores de Trombopoetina/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Colapso Estrutural
5.
Curr Opin Hematol ; 22(5): 445-51, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26154508

RESUMO

PURPOSE OF REVIEW: The human body produces and removes 10 platelets daily to maintain a normal steady-state platelet count. Platelet production must be tightly regulated to avoid spontaneous bleeding or arterial occlusion and organ damage. Multifaceted and complex mechanisms control platelet removal and production in physiological and pathological conditions. This review will focus on different mechanisms of platelet clearance, with focus on the biological significance of platelet glycans. RECENT FINDINGS: The Ashwell-Morrell receptor (AMR) recognizes senescent, desialylated platelets under steady state conditions. Desialylated platelets and the AMR are the physiological ligand-receptor pair regulating hepatic thrombopoietin (TPO) mRNA production, resolving the longstanding mystery of steady state TPO regulation. The AMR-mediated removal of desialylated platelets regulates TPO synthesis in the liver by recruiting JAK2 and STAT3 to increase thrombopoiesis. SUMMARY: Inhibition of TPO production downstream of the hepatic AMR-JAK2 signaling cascade could additionally contribute to the thrombocytopenia associated with JAK1/2 treatment, which is clinically used in myeloproliferative neoplasms.


Assuntos
Plaquetas/metabolismo , Trombopoese/fisiologia , Trombopoetina/metabolismo , Senescência Celular/fisiologia , Humanos , Janus Quinase 2/metabolismo , Fígado/metabolismo , Contagem de Plaquetas , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Trombocitopenia/fisiopatologia
6.
Blood ; 125(6): 1014-24, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25468568

RESUMO

Dynamins are highly conserved large GTPases (enzymes that hydrolyze guanosine triphosphate) involved in endocytosis and vesicle transport, and mutations in the ubiquitous and housekeeping dynamin 2 (DNM2) have been associated with thrombocytopenia in humans. To determine the role of DNM2 in thrombopoiesis, we generated Dnm2(fl/fl) Pf4-Cre mice specifically lacking DNM2 in the megakaryocyte (MK) lineage. Dnm2(fl/fl) Pf4-Cre mice had severe macrothrombocytopenia with moderately accelerated platelet clearance. Dnm2-null bone marrow MKs had altered demarcation membrane system formation in vivo due to defective endocytic pathway, and fetal liver-derived Dnm2-null MKs formed proplatelets poorly in vitro, showing that DNM2-dependent endocytosis plays a major role in MK membrane formation and thrombopoiesis. Endocytosis of the thrombopoietin receptor Mpl was impaired in Dnm2-null platelets, causing constitutive phosphorylation of the tyrosine kinase JAK2 and elevated circulating thrombopoietin levels. MK-specific DNM2 deletion severely disrupted bone marrow homeostasis, as reflected by marked expansion of hematopoietic stem and progenitor cells, MK hyperplasia, myelofibrosis, and consequent extramedullary hematopoiesis and splenomegaly. Taken together, our data demonstrate that unrestrained MK growth and proliferation results in rapid myelofibrosis and establishes a previously unrecognized role for DNM2-dependent endocytosis in megakaryopoiesis, thrombopoiesis, and bone marrow homeostasis.


Assuntos
Dinamina II/metabolismo , Endocitose , Megacariócitos/citologia , Trombopoese , Animais , Plaquetas/citologia , Plaquetas/metabolismo , Plaquetas/patologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Dinamina II/genética , Deleção de Genes , Megacariócitos/metabolismo , Megacariócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Trombopoetina/metabolismo , Transdução de Sinais , Esplenomegalia/genética , Esplenomegalia/metabolismo , Esplenomegalia/patologia , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patologia
7.
Thromb Haemost ; 110(2): 349-57, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23703656

RESUMO

Abacavir (ABC) has been associated with ischaemic cardiovascular events in HIV-infected patients, but the pathogenic mechanisms are unknown. Aim of our study was to assess whether ABC induces in vivo platelet activation and ex vivo platelet hyper-reactivity. In a retrospective, case-control study, in vivo platelet activation markers were measured in 69 HIV-infected patients, before starting therapy and after 6-12 months of either ABC (n=35) or tenofovir (TDF) (n=34), and compared with those from 20 untreated HIV-infected patients. A subgroup of patients was restudied after 28-34 months for ex vivo platelet reactivity. In vivo platelet activation markers were assessed by ELISA or flow cytometry, ex vivo platelet reactivity by light transmission aggregometry (LTA) and PFA-100®. Thein vitro effects of the ABC metabolite, carbovir triphosphate, on aggregation and intra-platelet cGMP were also studied. sPLA2, sPsel and sGPV increased significantly 6-12 months after the beginning of ABC, but not of TDF or of no treatment. Ex vivo platelet function studies showed enhanced LTA, shorter PFA-100® C/ADP closure time and enhanced platelet expression of P-sel and CD40L in the ABC group. The intake of ABC blunted the increase of intraplatelet cGMP induced by nitric oxide (NO) and acutely enhanced collagen-induced aggregation. Preincubation of control platelets with carbovir triphosphate in vitro enhanced platelet aggregation and blunted NO-induced cGMP elevation. In conclusion, treatment with ABC enhances in vivo platelet activation and induces platelet hyperreactivity by blunting the inhibitory effects of NO on platelets. These effects may lead to an increase of ischaemic cardiovascular events.


Assuntos
Fármacos Anti-HIV/efeitos adversos , Didesoxinucleosídeos/efeitos adversos , Infecções por HIV/sangue , Infecções por HIV/tratamento farmacológico , Ativação Plaquetária/efeitos dos fármacos , Adenina/efeitos adversos , Adenina/análogos & derivados , Adulto , Fármacos Anti-HIV/sangue , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Ligante de CD40 , Estudos de Casos e Controles , GMP Cíclico/sangue , Nucleotídeos de Desoxiguanina/efeitos adversos , Nucleotídeos de Desoxiguanina/sangue , Didesoxinucleosídeos/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/sangue , Organofosfonatos/efeitos adversos , Selectina-P/sangue , Fosfolipases A2 Secretórias/sangue , Agregação Plaquetária/efeitos dos fármacos , Estudos Retrospectivos , Tenofovir
8.
Am J Physiol Heart Circ Physiol ; 300(6): H2072-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21378140

RESUMO

Activated platelets express CD40L on their plasma membrane and release the soluble fragment sCD40L. The interaction between platelet surface CD40L and endothelial cell CD40 leads to the activation of endothelium contributing to atherothrombosis. Few studies have directly demonstrated an increased expression of platelet CD40L in conditions of in vivo platelet activation in humans, and no data are available on its relevance for endothelial activation. We aimed to assess whether platelets activated in vivo at a localized site of vascular injury in humans express CD40L and release sCD40L, whether the level of platelet CD40L expression attained in vivo is sufficient to induce endothelial activation, and whether platelet CD40L expression is inhibited by aspirin intake. We used the skin-bleeding-time test as a model to study the interaction between platelets and a damaged vessel wall by measuring CD40L in the blood emerging from a skin wound in vivo in healthy volunteers. In some experiments, shed blood was analyzed before and 1 h after the intake of 500 mg of aspirin. Platelets from the bleeding-time blood express CD40L and release soluble sCD40L, in a time-dependent way. In vivo platelet CD40L expression was mild but sufficient to induce VCAM-1 expression and IL-8 secretion in coincubation experiments with cultured human endothelial cells. Moreover, platelets recovered from the bleeding-time blood activated endothelial cells; an anti-CD40L antibody blocked this effect. On the contrary, the amount of sCD40L released by activated platelets at a localized site of vascular injury did not reach the concentrations required to induce endothelial cell activation. Soluble monocyte chemoattractant protein-1, a marker of endothelium activation, was increased in shed blood and correlated with platelet CD40L expression. Aspirin intake did not inhibit CD40L expression by platelets in vivo. We concluded that CD40L expressed by platelets in vivo in humans upon contact with a damaged vessel wall activates endothelium; aspirin treatment does not inhibit this mechanism.


Assuntos
Aspirina/farmacologia , Plaquetas/citologia , Plaquetas/metabolismo , Ligante de CD40/metabolismo , Comunicação Celular/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/lesões , Adulto , Aspirina/uso terapêutico , Aterosclerose/fisiopatologia , Aterosclerose/prevenção & controle , Plaquetas/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Endotélio Vascular/fisiopatologia , Feminino , Humanos , Inflamação/fisiopatologia , Interleucina-8/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Cardiovasculares , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Pele/lesões , Molécula 1 de Adesão de Célula Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA